EP3528849A1 - Nanocage - Google Patents

Nanocage

Info

Publication number
EP3528849A1
EP3528849A1 EP17790822.5A EP17790822A EP3528849A1 EP 3528849 A1 EP3528849 A1 EP 3528849A1 EP 17790822 A EP17790822 A EP 17790822A EP 3528849 A1 EP3528849 A1 EP 3528849A1
Authority
EP
European Patent Office
Prior art keywords
ferritin
variant
seq
nanocage
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP17790822.5A
Other languages
German (de)
English (en)
Inventor
Geoffrey Baldwin
James Field
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ip2ipo Innovations Ltd
Original Assignee
Imperial Innovations Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Imperial Innovations Ltd filed Critical Imperial Innovations Ltd
Publication of EP3528849A1 publication Critical patent/EP3528849A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0028Disruption, e.g. by heat or ultrasounds, sonophysical or sonochemical activation, e.g. thermosensitive or heat-sensitive liposomes, disruption of calculi with a medicinal preparation and ultrasounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6923Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being an inorganic particle, e.g. ceramic particles, silica particles, ferrite or synsorb
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6925Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a microcapsule, nanocapsule, microbubble or nanobubble
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0045Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent agent being a peptide or protein used for imaging or diagnosis in vivo
    • A61K49/0047Green fluorescent protein [GFP]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0063Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres
    • A61K49/0069Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form
    • A61K49/0089Particulate, powder, adsorbate, bead, sphere
    • A61K49/0091Microparticle, microcapsule, microbubble, microsphere, microbead, i.e. having a size or diameter higher or equal to 1 micrometer
    • A61K49/0093Nanoparticle, nanocapsule, nanobubble, nanosphere, nanobead, i.e. having a size or diameter smaller than 1 micrometer, e.g. polymeric nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5169Proteins, e.g. albumin, gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/24Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • C07K14/245Escherichia (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/84Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving inorganic compounds or pH
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • the present invention relates to nanocages, and in particular to protein nanocages, and especially ferritin nanocages.
  • the invention extends to variant ferritin polypeptides and their encoding nucleic acids, mutant ferritin nanocages, and their uses in diagnostics and drug delivery, as well as in phenotypic screens in drug development.
  • Protein nanocages are a class of protein that self-assemble to form a three dimensional structure with a central cavity.
  • Ferritin is one such protein, it is found in all kingdoms of life and naturally acts to store iron and so protect the host from oxidative damage caused by the Fenton reaction. Ferritins have received a significant amount of attention for their potential bionanotechnology applications 1 .
  • ferritin nanocages have an external diameter of 12 nm and an internal cavity of 8 nm. It has been demonstrated that ferritin nanocages can be reversibly disassembled by a shift in pH 2 and this has been used to encapsulate the anti-cancer drug doxorubicin (Dox) at a ratio of approximately five Dox molecules per cages, while this approach has been useful, it suffers from the problem of poor efficiency, as typically only 50% or less of fully assembled cages are recovered 3,4 .
  • Dox anti-cancer drug doxorubicin
  • the proportion of the active agent, Dox, that can be encapsulated into the ferritin using a passive encapsulation technique, where the nanocage is reformed in the presence of the drug is only around 0.1 to 0.4 % 3 , which is very low and therefore wasteful in terms of drug loading.
  • chimeric ferritin molecules can be made by linking different peptides to the N-terminus of the protein. By mixing the two types of ferritin m vitro and disassembling and reassembling using a pH switch, different peptides can be incorporated onto the same nanocage structure. This provides an interesting method by which multi-valent epitopes may be attached to the nanocage, but with limited control of the distribution 11 .
  • Nanoparticles for the targeted delivery of drugs in vivo is an attractive idea that has been the subject of significant research. Nevertheless, over the last 10 years it has not been possible to significantly improve the targeting ratio of the designed
  • nanoparticles 12 Most of the nanoparticles studied have been chemically based in a size range of 10-200 nm and rely on the enhanced permeability and retention effect (EPR) associated with many tumours.
  • EPR enhanced permeability and retention effect
  • MPS mononuclear phagocytic system
  • EPR enhanced permeability and retention effect
  • Ferritin presents an attractive alternative to many chemical-based agents. It is large enough to be retained in the circulation (>8 nm), but is also biocompatible and non- immunogenic 4 ' 11 . It is also small enough that it will have better tumour penetrating properties, since size ( ⁇ 50 nm) is an important factor in targeting efficiency 12 . In addition, it has been proposed that invasion of ferritin to a tumour may occur via an intra-cell transport mechanism 13,14 and so will not be entirely dependent on the EPR effect for tumour invasion.
  • ferritin nanocages and components thereof which can be used for targeted delivery of drugs to cells in vitro and in vivo and/or diagnosis, and in phenotypic screens in drug development.
  • the inventors set out to engineer a biocompatible platform that will facilitate a modular and generic approach.
  • the inventors have developed a variant ferritin polypeptide in which the dimeric subunit interface has been mutated such that it is unable to self-assemble to form a nanocage structure.
  • the mutant upon contacting the variant ferritin with a nucleating metallic core (such as a gold nanoparticle), the mutant self-assembles around the core, thereby forming a nanocage encapsulating the core. Furthermore, it is possible to encapsulate active agents, such as small molecule drugs, into the self-assembling nanocage structure, by attaching the active agent to the metal core prior to contacting it with the variant ferritin polypeptide.
  • active agents such as small molecule drugs
  • the invention thus provides a novel mechanism for the encapsulation of drugs into the ferritin nanocage without harsh denaturation conditions that are used in known systems.
  • the inventors have also shown that the variant nanocage can be modified to be fluorescent by fusion of an N-terminal fluorescent protein to the mutant ferritin, for use in diagnostics and imaging experiments.
  • the nanocage can be specifically bound to antibodies or antigen-binding fragments thereof, and targeted to cells by further fusion of an antibody binding domain to the N- terminus of the variant ferritin, so that antibody-bound protein can specifically bind to target cells.
  • this antibody-based targeting platform can be used for the targeted delivery of drugs into cells, for example tumour cells.
  • a variant ferritin polypeptide comprising a modified amino acid sequence of a wild-type ferritin polypeptide, the modified sequence being in a dimeric subunit interface or the N-terminus of the polypeptide, wherein the variant is incapable of assembling into a ferritin nanocage unless it is contacted with a nucleating agent.
  • the variant ferritin of the invention is biocompatible and not immunogenic.
  • the inventors have engineered several embodiments of ferritin polypeptide monomers, which only self-assemble into a nanocage in the presence of a nucleating agent. These modified nanocage monomers can be used in diagnosis or in therapy, such as to facilitate the delivery of drugs into cells, either in vivo or in vitro.
  • the variant ferritin polypeptide comprises a modified bacterial ferritin, also known as bacterioferritin (Bfr).
  • the bacterioferritin may be isolated from E. coli. It contains 24 subunits and 12 heme groups that bind between the dimeric protein interface.
  • the nucleic acid (SEQ ID No:i) and amino acid (SEQ ID No:2) sequences of wild-type E.coli bacterioferritin are known, and may be represented herein as SEQ ID No:i and SEQ ID No: 2, or a fragment or variant thereof, as follows:-
  • the variant bacterioferritin comprises a His tag.
  • the His tag is encoded by a nucleic acid sequence (SEQ ID No:3) or comprises an amino acid sequence (SEQ ID No:4), or a fragment of variant thereof, substantially as set out in SEQ ID No:3 and SEQ ID No:4, as follows:-
  • the variant bacterioferritin comprises an N-terminal His tag.
  • the variant bacterioferritin is preferably encoded by a nucleic acid (SEQ ID No:s) or comprises an amino acid (SEQ ID No:6) sequence, or fragment of variant thereof, substantially as set out in SEQ ID No: 5 and SEQ ID No:6, as follows:
  • the variant bacterioferritin comprises an amino acid sequence configured to bind a nucleating agent, and may for example be a silica binding peptide, or a metal binding peptide, such as gold, copper, iron.
  • the variant may comprise a gadolinium binding peptide.
  • the variant bacterioferritin comprises a gold-binding peptide.
  • a suitable metal binding peptide maybe encoded by a nucleic acid sequence (SEQ ID No:7) or comprises an amino acid sequence (SEQ ID No:8), or a fragment of variant thereof, substantially as set out in SEQ ID No:7 and SEQ ID No:8, as follows:-
  • the nucleating agent binding peptide is a Oterminal nucleating agent binding peptide.
  • the variant bacterioferritin is preferably encoded by a nucleic acid sequence (SEQ ID No: 9) or comprises an amino acid sequence (SEQ ID No: 10), or a fragment or variant thereof, substantially as set out in SEQ ID No:9 or SEQ ID No:io, as follows:
  • the variant bacterioferritin may comprise an N- terminal His tag and a C-terminal nucleating agent binding peptide.
  • the variant bacterioferritin is encoded by a nucleic acid sequence (SEQ ID No:ii) or comprises an amino acid sequence (SEQ ID No: 12), or a fragment or variant thereof, substantially as set out in SEQ ID No: 11 or SEQ ID No:i2, as follows:
  • the inventors were surprised to observe that the addition of the N-terminal His-tag meant that the bacterioferritin did not dimerise or purify in its nanocage composition, but instead as individual monomers.
  • the variant bacterioferritin surprisingly formed a higher order structure consistent with a nanocage being formed around the gold nanoparticle.
  • the subtle modification of the bacterioferritin sequence with an N- terminal His tag has destabilised the nanocage structure of bacterioferritin under normal physiological conditions, and the use of a C-terminal metal binding peptide is sufficient to establish metal binding peptide-templated assembly of a nanocage without using harsh denaturation conditions.
  • the bacterioferritin is expressed in a bacterial host using a construct comprising a promoter, a ribosomal binding site (RBS) and nucleic acid encoding a His tag.
  • the promoter used in the construct maybe a compound promoter with the constitutive J23100 promoter in combination with the inducible T7 promoter.
  • the nucleic acid (SEQ ID No:i3) and amino acid (SEQ ID No:i4) sequences of a preferred bacterial expression construct maybe represented herein as SEQ ID No:i3 and SEQ ID No:i4, respectively, or a fragment or variant thereof, as follows:-
  • the variant ferritin polypeptide comprises a modified mammalian ferritin, and most preferably modified human ferritin.
  • the variant human ferritin comprises one or more modification that disrupts the dimeric subunit interface of the wild-type human polypeptide, thereby rendering the variant incapable of forming heavy chain dimers unless it is contacted with a nucleating agent.
  • Human ferritin may be composed of the light chain ferritin subunit (1FTN) or heavy chain ferritin subunit (hFTN), or a combination of both. By expressing either 1FTN or hFTN in a host (e.g. E. coli), it is possible to create ferritin variant nanocages that consist of only a single protein monomer.
  • nucleic acid SEQ ID No:i5 and amino acid (SEQ ID No:i6) sequences of wild- type human heavy chain ferritin are known, and may be represented herein as SEQ ID No:i5 and SEQ ID No:i6, or a fragment or variant thereof, substantially as foUows:-
  • nucleic acid SEQ ID No:i7 and amino acid (SEQ ID No:i8) sequences of wild- type human light chain ferritin are known, and may be represented herein as SEQ ID No:i7 and SEQ ID No:i8, or a fragment or variant thereof, substantially as follows:-
  • the inventors analysed over 147 conserved ferritin proteins, and managed to surprisingly identify several evolutionarily conserved domains at the dimeric interface of human ferritin proteins (heavy and light chains) that contain at least one hydrophobic residue (see Table l in Example 2). Hydrophobic residues within these conserved motifs were then carefully selected for site specific mutagenesis (see Figures 4C and 4D). Four mutations were created in the heavy chain variant of ferritin [hFTN (L29A L36A I81A L83A)] and four mutations were also made in the light chain variant of the polypeptide QFT (L32A F36A L67A F79A)] according to the conserved motifs that were identified.
  • the variant ferritin polypeptide comprises a variant human heavy chain ferritin.
  • the variant human heavy chain ferritin comprises one or more modification that disrupts the dimeric subunit interface of the wild-type polypeptide, thereby rendering the variant incapable of forming heavy chain dimers unless it is contacted with a nucleating agent
  • the variant human heavy chain ferritin comprises one or more modification in the wild-type polypeptide, wherein one or more hydrophobic residue in the heavy chain dimeric subunit interface of the polypeptide is substituted with a small amino acid residue, thereby rendering the variant incapable of forming heavy chain dimers, and hence higher order nanocages, unless it is contacted with a nucleating agent.
  • the heavy chain dimeric subunit interface comprises or consists of amino acid residues as set out in Table 1, i.e. SEQ ID No's: 19, 20, 21, 22 and 29.
  • the variant heavy chain ferritin polypeptide comprises at least one modification in amino acids 29, 36, 81 or 83 of SEQ ID No: 16.
  • the variant heavy chain ferritin polypeptide comprises at least two, more preferably at least three, and most preferably four modifications in amino acids 29, 36, 81 or 83 of SEQ ID No: 16.
  • the variant heavy chain ferritin polypeptide is formed by
  • the modification at amino acid L29 comprises a substitution with an alanine, i.e. L29A.
  • the modification at amino acid L36 comprises a substitution with an alanine, i.e. L36A.
  • the modification at amino acid I81 comprises a substitution with an alanine, i.e. I81A.
  • the modification at amino acid L83 comprises a substitution with an alanine, i.e.
  • the variant human heavy chain ferritin polypeptide (L29A L36A I81A L83A) is encoded by a nucleic acid (SEQ ID No:30) or comprises an amino acid (SEQ ID No:3i) sequence, or fragment of variant thereof, substantially as set out in SEQ ID No: 30 and SEQ ID No:3i, as follows:
  • the variant ferritin polypeptide comprises a variant human light chain ferritin.
  • the variant human light chain ferritin comprises one or more modification that disrupts the dimeric subunit interface of the wild-type polypeptide, thereby rendering the variant incapable of forming light chain dimers unless it is contacted with a nucleating agent
  • the or each modification comprises substituting one or more hydrophobic residue in the light chain dimeric subunit interface of the polypeptide with a small amino acid residue, thereby rendering the variant incapable of forming light chain dimers and hence higher order nanocages, unless it is contacted with a nucleating agent.
  • the light chain dimeric subunit interface comprises or consists of amino acid residues as set out in Table 1, i.e. SEQ ID No's: 23, 24, 25, 26, 27, 28, and 29.
  • the variant light chain ferritin polypeptide comprises at least one modification in amino acids 32, 36, 67 or 79 of SEQ ID No:i8.
  • the variant light chain ferritin polypeptide comprises at least two, more preferably at least three, and most preferably four modifications in amino acids 32, 36, 67 or 79 of SEQ ID No: 18.
  • the variant light chain ferritin polypeptide is formed by modification of amino acid residue L32, F36, L67 and/or F79 of SEQ ID No:i8.
  • the modification at amino acid L32 comprises a substitution with an alanine, i.e. L32A.
  • the modification at amino acid F36 comprises a substitution with an alanine, i.e. F36A.
  • the modification at amino acid L67 comprises a substitution with an alanine, i.e. L67A.
  • the modification at amino acid F79 comprises a substitution with an alanine, i.e. F79A.
  • the variant human light chain ferritin (L32A F36A L67A F79A) is encoded by a nucleic acid (SEQ ID No:32) or comprises an amino acid (SEQ ID No:33) sequence, or a fragment or variant thereof, substantially as set out in SEQ ID No: 32 and SEQ ID No:33, as follows:
  • the variant ferritin which maybe bacterial ferritin or human ferritin (heavy or light chain), comprises a fluorophore, such as green fluorescent protein (GFP), red fluorescent protein (RFP) or cyan fluorescent protein (CFP).
  • GFP green fluorescent protein
  • RFP red fluorescent protein
  • CFP cyan fluorescent protein
  • a preferred fluorophore comprises GFP, the nucleic acid (SEQ ID No:34) and amino acid (SEQ ID No:35) sequences of which are known, and are substantially as set out in SEQ ID No: 34 and SEQ ID No:35, as follows:
  • the fluorophore is preferably disposed at or towards the N-terminus of the variant ferritin.
  • the variant human heavy chain ferritin is encoded by a nucleic acid (SEQ ID No:36) or comprises an amino acid (SEQ ID No:37) sequence, or a fragment of variant thereof, substantially as set out in SEQ ID No: 36 and SEQ ID No:37, as follows:
  • the variant human light chain ferritin is encoded by a nucleic acid (SEQ ID No:38) or comprises an amino acid (SEQ ID No:39) sequence, or fragment of variant thereof, substantially as set out in SEQ ID No: 38 and SEQ ID No:39, as follows:
  • the variant human heavy or light chain ferritin comprises a His tag, more preferably an N-terminal His tag.
  • the His tag is encoded by a nucleic acid sequence (SEQ ID No:3) or comprises an amino acid sequence (SEQ ID No:4), or a fragment of variant thereof, as disclosed herein.
  • the variant human heavy chain ferritin is encoded by a nucleic acid (SEQ ID No:40) or comprises an amino acid (SEQ ID No:4i) sequence, or a fragment of variant thereof, substantially as set out in SEQ ID No: 40 and SEQ ID No:4i, as follows:
  • the variant human light chain ferritin is encoded by a nucleic acid (SEQ ID No:42) or comprises an amino acid (SEQ ID No:43) sequence, or a fragment of variant thereof, substantially as set out in SEQ ID No: 42 and SEQ ID No:43, as follows:
  • the variant human heavy or light chain ferritin comprises a nucleating agent binding peptide, for example a silica binding peptide, or a metal binding peptide, such as gold, copper, iron, or it maybe a gadolinium binding peptide.
  • the variant human heavy or light chain ferritin comprises a gold-binding peptide.
  • a suitable metal binding peptide may comprise or consist of an amino acid sequence substantially as set out in SEQ ID No:8, or a fragment of variant thereof, or encoded by a nucleic acid sequence substantially as set out in SEQ ID No: 7.
  • the nucleating agent binding peptide is a C-terminal nucleating agent binding peptide.
  • the variant human heavy or light chain ferritin comprises an N- terminal His tag and a C-terminal nucleating agent binding peptide.
  • the variant human heavy chain ferritin is encoded by a nucleic acid (SEQ ID No:44) or comprises an amino acid (SEQ ID No:45) sequence, or a fragment or variant thereof, substantially as set out in SEQ ID No: 44 and SEQ ID No:45, as follows:
  • the variant human light chain ferritin is encoded by a nucleic acid (SEQ ID No:46) or comprises an amino acid (SEQ ID No:47) sequence, or fragment or variant thereof, substantially as set out in SEQ ID No: 46 and SEQ ID No:47, as follows:
  • the variant ferritin which maybe bacterial or human ferritin (which maybe the heavy or light chain), comprises an amino acid sequence configured to bind to an antibody or antigen binding fragment thereof, such as an IgG isotype antibody.
  • a preferred antibody or antigen binding fragment thereof binding amino acid sequence comprises a Z-domain, which is a derivative of Staphylococcus protein A, and which is an engineered version of the IgG binding domain of protein A with greater stability and a higher binding af inity for the Fc antibody domain.
  • the Z domain sequence may be encoded as a single domain, it is preferably coded as a repeat so that two tandem domains are disposed adjacent to one another (i.e. ZZ), preferably with suf icient redundancy in the DNA code such that the sequences are not direct repeats.
  • the nucleic acid (SEQ ID No:48) and amino sequences (SEQ ID No:49) of ZZ are known, and are as set out in SEQ ID No: 48 and SEQ ID No:49, as follows:
  • the antibody or antigen binding fragment thereof binding peptide is provided at or towards the N-terminus of the variant ferritin polypeptide.
  • the variant human heavy chain ferritin is encoded by a nucleic acid (SEQ ID No:50) or comprises an amino acid (SEQ ID No:5i) sequence, or fragment or variant thereof, substantially as set out in SEQ ID No: 50 and SEQ ID No:5i, as follows:
  • the variant bacterioferritin is encoded by a nucleic acid (SEQ ID No:52) or comprises an amino acid (SEQ ID No:53) sequence, or fragment or variant thereof, substantially as set out in SEQ ID No: 52 and SEQ ID No:53, as follows:
  • the inventors have also constructed a comprehensive series of fusion proteins which comprise the wild-type ferritin polypeptide (i.e. bacterial, or human light chain, or human heavy chain) fused to one or more amino acid sequence of a His tag, a nucleating agent binding peptide, GFP (i.e. fluorophore) and/or an antibody binding peptide.
  • wild-type ferritin polypeptide i.e. bacterial, or human light chain, or human heavy chain
  • GFP i.e. fluorophore
  • a fusion protein comprising wild-type ferritin and one or more peptide selected from a group consisting of: an antibody or antigen binding fragment thereof binding peptide; a fluorophore; a His tag; and a nucleating agent binding peptide.
  • the fusion protein may comprise various combinations of the fluorophore, His tag, nucleating agent binding peptide, and antibody binding peptide, i.e. some or all of these features.
  • the fusion protein comprises bacterioferritin, more preferably comprising or consisting of an amino acid sequence substantially set out as SEQ ID No: 2, or is encoded by a nucleic acid sequence substantially set out as SEQ ID No:i, or fragments or variants thereof.
  • the fusion protein comprises human ferritin, which maybe light chain or heavy chain ferritin.
  • the fusion protein comprises or consists of an amino acid sequence substantially set out as SEQ ID No: 16 or 18, or is encoded by a nucleic acid sequence substantially set out as SEQ ID No:i5 or 17, or fragments or variants thereof.
  • the fluorophore comprises GFP.
  • GFP may comprise or consist of an amino acid sequence substantially set out as SEQ ID No: 35, or is encoded by a nucleic acid sequence substantially set out as SEQ ID No:34, or fragments or variants thereof.
  • the fluorophore is disposed at or towards the N-terminus of the variant ferritin.
  • the His tag comprises or consists of an amino acid sequence substantially set out as SEQ ID No: 4, or is encoded by a nucleic acid sequence substantially set out as SEQ ID No:3, or fragments or variants thereof.
  • the His tag is disposed at or towards the N-terminus of the variant ferritin.
  • the nucleating agent binding peptide comprises a silica binding peptide, or a metal binding peptide, such as gold, copper, or iron.
  • the nucleating agent binding peptide comprises a gold-binding peptide.
  • the gold-binding peptide comprises or consists of an amino acid sequence substantially as set out in SEQ ID No:8, or is encoded by a nucleic acid sequence substantially set out as SEQ ID No:7, or fragments or variants thereof.
  • the antibody or antigen binding fragment thereof binding peptide comprises a repeated Z-domain.
  • the repeated Z domain comprises or consists of an amino acid sequence substantially set out as SEQ ID No: 49, or is encoded by a nucleic acid sequence substantially set out as SEQ ID No: 48, or fragments or variants thereof.
  • the fusion protein comprises wild-type heavy chain human ferritin, GFP and a His tag.
  • the fusion protein of the second aspect is encoded by a nucleic acid substantially as set out in SEQ ID No: ⁇ 54, or comprises an amino acid substantially as set out in SEQ ID No:55, or fragments or variants thereof.
  • the fusion protein comprises wild-type light chain human ferritin, GFP and a His tag.
  • the fusion protein of the second aspect is encoded by a nucleic acid substantially as set out in SEQ ID No:s6, or comprises an amino acid substantially as set out in SEQ ID No:57, or fragments or variants thereof.
  • the fusion protein comprises wild-type heavy chain human ferritin, GFP, a His tag and a nucleating agent binding peptide, which is preferably a metal (e.g. gold) binding peptide.
  • the fusion protein of the second aspect is encoded by a nucleic acid substantially as set out in SEQ ID No:s8, or comprises an amino acid substantially as set out in SEQ ID No:59, or fragments or variants thereof.
  • the fusion protein comprises wild-type light chain human ferritin, GFP, a His tag and a nucleating agent binding peptide, which is preferably a metal (e.g. gold) binding peptide.
  • the fusion protein of the second aspect is encoded by a nucleic acid substantially as set out in SEQ ID No: 60, or comprises an amino acid substantially as set out in SEQ ID No:6i, or fragments or variants thereof.
  • the fusion protein comprises wild-type heavy chain human ferritin, GFP, a His tag, and an antibody or antigen binding fragment thereof binding peptide.
  • the fusion protein of the second aspect is encoded by a nucleic acid substantially as set out in SEQ ID No:62, or comprises an amino acid substantially as set out in SEQ ID No:63, or fragments or variants thereof.
  • Preferred peptide linker sequences used between open reading frames in the above variant and wild type ferritin polypeptides and fusion proteins include:
  • fusion protein was created that lacked the GFP so that cell delivery could be performed with phenotypic cell assays using a Vybrant cell staining kit without interfering fluorescence signals arising from GFP.
  • Variant ferritin fusions were created with different linker amino acid sequences.
  • these fusion proteins are preferably encoded by a nucleic acid substantially as set out in SEQ ID No:72, 74 and 76, or may comprise an amino acid substantially as set out in SEQ ID No:73, 75 and 77, or fragments or variants thereof.
  • the variant ferritin polypeptides developed by the inventors have been mutated in such a way that they cannot self-assemble to form a nanocage unless they have been contacted with a nucleating agent, such as a metallic (e.g. gold) nanoparticle, in which case the mutant self-assembles around the metallic core, thereby forming a nanocage and encapsulating the core.
  • a nucleating agent such as a metallic (e.g. gold) nanoparticle
  • an isolated nucleic acid comprising or consisting of a nucleotide sequence encoding the variant ferritin polypeptide of the first aspect or the fusion protein of the second aspect, or a fragment or variant thereof.
  • the nucleic acid preferably comprises or consists of one or more of the nucleotide sequences described herein.
  • Preferred nucleic acids comprise or consist of a nucleotide sequence substantially as set out in any one of SEQ ID No: 5, 9, 11, 30, 32, 36, 38, 40, 42, 44, 46, 50, 52, 54, 56, 58, 60 or 62.
  • a ferritin nanocage comprising the variant ferritin polypeptide of the first aspect or the fusion protein of the second aspect, and a nucleating agent.
  • the nanocage may comprise a plurality of identical monomers of ferritin polypeptide or fusion protein.
  • each monomer may comprise ferritin, and one or more domain selected from a group consisting of: an antibody or antigen binding fragment thereof binding peptide; a fluorophore; a His tag; and a nucleating agent binding peptide.
  • the monomer comprises human ferritin, optionally the light chain or heavy chain ferritin.
  • the monomer may comprise His-ZZ-hFtn(L29A L36A L81A L83A).
  • the resultant nanocage will contain the ZZ domain and the GFP domain on each subunit.
  • the nanocage may comprise a plurality of different monomers of ferritin polypeptide or fusion protein.
  • the nanocage may comprise first and second monomers comprising ferritin, and one or more domain selected from a group consisting of: an antibody or antigen binding fragment thereof binding peptide; a fluorophore; a His tag; and a nucleating agent binding peptide, wherein the first and second monomers have different combinations of domains.
  • the monomer comprises human ferritin, optionally the light chain or heavy chain ferritin.
  • compound or mixed nanocages composed of different types of ferritin subunit were also created.
  • a first monomer may comprise His-ZZ-hFtn(L2gA L36A L81A L83A) and a second monomer may comprise His-GFP-hFtn(L29A L36A L81A L83A). Since the hFtn part of the resultant fusion protein is identical, nanocages form that contain the ZZ domain on some subunits, and the GFP domain on others.
  • a method of preparing a ferritin nanocage comprising contacting the variant ferritin polypeptide of the first aspect or the fusion protein of the second aspect, with a nucleating agent.
  • the nucleating agent preferably comprises a nanoparticle having an average diameter of about i-50onm, more preferably l-ioonm, even more preferably 2-5onm, and most preferably 3-ionm.
  • the nucleating agent is metallic.
  • the nucleating agent maybe gold, iron, or copper.
  • the agent may comprise a gadolinium binding peptide.
  • the ferritin polypeptide encapsulates the nucleating agent.
  • the ferritin nanocage encapsulates a gold nanoparticle.
  • the method according to the invention can be used to easily create a ferritin nanocage. Furthermore, the method according to the invention does not require the use of harsh denaturation conditions in order to create a nanocage, which is advantageous because it reduces the likelihood of destroying the integrity of the reformed nanocage.
  • the nanocage can be modified to be fluorescent by fusion of an N-terminal fluorescent protein to the ferritin monomer, for use in diagnostics and imaging experiments.
  • the ferritin nanocage is functionalised with an imaging agent, such as a fluorescent protein or fluorophore.
  • the nanocages of the invention can be modified to become fluorescent by fusion or conjugation of a fluorescent protein, for example GFP or the like.
  • the fluorescent protein is fused at or towards the N-terminus of the ferritin polypeptide.
  • nanocage can be any nanocage. Furthermore, the inventors have also demonstrated that the nanocage can be any nanocage.
  • the nanocage of the invention has been successfully functionalised with anti-EGFR antibodies.
  • the antibody binding domain is fused to the N-terminus of the ferritin monomer.
  • specific targeting and endocytosis of the nanocage can be achieved by modifying the ferritin with an IgG binding domain. This enables the nanocage to bind to IgG type antibodies in a simple binding reaction. Thus, binding of the ferritin nanocage to an antibody leads to specific targeting of cells.
  • an antibody that targets endocytic receptors such as the EGFR receptor
  • the nanocage can be endocytosed (Goh & Sorkin, CSH Perspect. Biol. 5(5), 2013), which leads to delivery of the nanocage and its contents directly into the cell. As described in Examples 11 and 12, the nanocage of the invention has been successfully functionalised with anti-EGFR antibodies.
  • the ferritin nanocage comprises, or is functionalised with an antibody or antigen binding fragment thereof.
  • the antibody or antigen binding fragment thereof is immunospecific for endocytic receptors.
  • the nanocage is endocytosed leading to delivery of the nanocage and its contents directly into the target cell.
  • a preferred antibody or antigen binding fragment thereof binding amino acid sequence comprises a Z-domain, which is a derivative of Staphylococcus protein A. This is an engineered version of the IgG binding domain of protein A with greater stability and a higher binding affinity for the Fc antibody domain.
  • the ferritin nanocage is functionalised with an IgG antibody.
  • the ferritin nanocage is functionalised by binding to the Fc domain of the antibody, so that antigen recognition is not compromised through direct interaction with the Fv domain.
  • the antibody or antigen binding fragment thereof preferably exhibits immunospecificity for a target cell or tissue.
  • the nanocage can be targeted to specific cells (e.g. a tumour cell) by fusion of an antibody binding domain at or towards the N-terminus of the ferritin polypeptide.
  • functionalised nanocages according to the invention can be targeted to specific cells, and simultaneously visualised.
  • the nanocage of the invention can be used as a vector for delivering drug molecules to a target cell or tissue.
  • a ferritin nanocage according to invention for use as a vector for the delivery of a payload molecule, preferably a drug molecule, to a target biological environment.
  • the nucleating agent which is preferably a metallic nanoparticle, maybe bound to a payload which maybe an active agent, such as a drug molecule.
  • a payload which maybe an active agent, such as a drug molecule.
  • the ferritin nanocage is configured, in use, to encapsulate and carry the payload molecule to a target biological environment.
  • the nanocage comprises an internal cavity in which the payload molecule is contained, wherein the payload molecule is capable of being active when the nanocage is at least adjacent to the target biological environment.
  • a method of encapsulating a payload molecule preferably a drug molecule, in a ferritin nanocage, the method comprising contacting the variant ferritin polypeptide of the first aspect or the fusion protein of the second aspect with a nucleating agent conjugated to a payload molecule and allowing the polypeptide or protein to self-assemble into a nanocage, thereby encapsulating the payload molecule.
  • the payload molecule described herein may be an active agent, such as a small molecule drug, which may be bound to the nucleating agent prior to encapsulation and subsequent mixing of the variant ferritin polypeptide or fusion protein.
  • the molecular weight of the payload molecule may be 50 Da to 10 kDa, preferably 100 Da to lkDa, more preferably 250 Da to 1000 Da.
  • the anti-cancer drug doxorubicin was used as an exemplary active agent in the
  • Another preferred payload molecule is paclitaxel, as described in Example 11.
  • the payload molecule maybe an antibiotic, such as actinomycin, as described in Example 12.
  • the payload molecule may therefore be a peptide, or cyclic peptide.
  • Yet another preferred payload molecule is actinomycin-D. As described in Example 14, using mass spectrometry, 13.3 actinomycin D molecules have been encapsulated by the nanocage.
  • the payload molecule may be bound or conjugated to the nucleating agent by van der Waal's forces or ionic forces.
  • the nucleating agent-drug conjugate leads to the formation of the ferritin nanocage which encapsulates the nucleating agent and the active agent conjugates thereto within the nanocage.
  • the method according to the invention can be used to easily load a drug into a ferritin nanocage.
  • a further advantage of the invention is that it can be used to widen the therapeutic window of drugs that are otherwise incapable of permeating cells without assistance.
  • the nucleating agent is a metallic nanoparticle, more preferably a gold nanoparticle.
  • the inventors have generated an innovative approach to producing and using ferritin as a targetable drug delivery agent. They have engineered mutations in the ferritin monomer so that it does not form a nanocage in isolation, and can be purified in its monomelic state. When mixed with a metallic nanoparticle, the nanoparticle acts as a nucleation site and the nanocage specifically reforms around the metallic nanoparticle. Functionalising the nanocage with a suitable antibody ensures that the nanocage is targeted to a target site.
  • Example 5 explains how the nanocage can be targeted to
  • MNK1.1 mouse natural killer cells
  • HT29 colonal cancer cell lines
  • a method of targeting a ferritin nanocage to a target biological environment comprising functionalising the ferritin nanocage of the third aspect with an antibody or antigen binding fragment thereof which is immunospecific for a target cell, and allowing the functionalised nanocage to be targeted to the target biological environment.
  • the ability to target ferritin nanocages to specific cell types via the binding of antibodies creates huge possibilities for the diagnosis and treatment of disease.
  • the ferritin nanocage reaches the desired target biological environment, it is subjected to a decrease in pH associated with lysosomes, which causes the otherwise
  • the nanocages can be made fluorescent, they can be used in imaging methods to identify specific cell types displaying known epitope disease markers. This creates possibilities for their use in the diagnosis of cancer types in imaging accessible locations.
  • the target biological environment may be a cell or tissue, such as a cancer or tumour cell. Examples are cancers accessible via Gl-tract, such as oesophageal, stomach, colorectal, liver, pancreatic, gall bladder.
  • cancers near to the surface of the body would be accessible for diagnosis including skin cancer and neck and throat cancers.
  • the drug-encapsulated complex contains a metallic (e.g. gold) nanoparticle
  • a mechanism for the activated release of drugs is also possible.
  • Gold nanoparticles absorb light due to their plasmonic effect and laser irradiation maybe used to cause localised heating of the nanoparticle proportional to the intensity of the incident laser irradiation.
  • laser induced heating may therefore be used to activate the release of the encapsulated drug, since localised heating will lead to the thermal disassembly of the nanocage complex in the same way that the pH drop associated with endosomes does.
  • This type of approach can make use of current endoscope technology that can both locally deliver compounds, image and treat using laser light sources. The inventors therefore consider that this type of nanocage device would fit with current therapeutic practices and approaches.
  • the ability to encapsulate drugs into the nanocage also provides the possibility of combined diagnostic and therapy (theranostic) approaches.
  • the variant ferritin polypeptide of the first aspect, the fusion protein of the second aspect or the ferritin nanocage of the third aspect for use in therapy or diagnosis.
  • the variant ferritin polypeptide of the first aspect, the fusion protein of the second aspect or the ferritin nanocage of the third aspect for use in the treatment, prevention or amelioration of disease, preferably cancer.
  • a method of treating, ameliorating or preventing a disease, preferably cancer comprising administering, to a subject in need of such treatment, a therapeutically effective amount of the variant ferritin polypeptide of the first aspect, the fusion protein of the second aspect or the ferritin nanocage of the third aspect.
  • the method comprises administering the ferritin nanocage of the third aspect to the subject, and then exposing the nanocage to heat such that it disassembles, thereby releasing the payload molecule.
  • the heat may be provided by a suitable heat source, such as a laser.
  • a suitable heat source such as a laser.
  • the principle of laser-induced drug release has been demonstrated by examining the fluorescence polarisation of a fluorescently-bound molecule within the nanocage, such as Dox. Anisotropy provides an intensity independent measure of the degree of polarisation within a sample. When a fluorescent molecule absorbs plane polarised light, it will be emitted in the same plane as the excitation source. However, during the fluorescence lifetime, between absorption and emission, the molecule may rotate.
  • the emitted light will be relative to the new orientation of the molecule.
  • a fluorescent molecule encapsulated in the nanocage will therefore have a very high anisotropy value. Laser irradiation of the metallic nanoparticle leads to the breakdown of the nanocage and release of a fluorescent compound, and this can be imaged by a significant reduction in the measured anisotropy.
  • a heat source to heat a ferritin nanocage according to the third aspect comprising an encapsulated payload molecule, to disassemble the nanocage and thereby release the payload molecule.
  • the heat source maybe a laser.
  • the inventors also believe that the nanocage can be used in phenotypic screens for use in drug development.
  • a phenotypic assay comprising the ferritin nanocage according to the third aspect.
  • the inventors have demonstrated the ability to use the ferritin nanocage as a platform technology for the delivery of small molecule drugs into cells. Because the technology provides a defined process for the encapsulation and assembly of the nanocage complex, it can be envisioned as a generic method for the delivery of compounds into cells. The binding of small molecule compounds to the metallic nanoparticle core would work for a wide variety of ionic, electrostatic and hydrophobic interactions. The assembly of the mutant nanocage around the drug-bound
  • nanoparticle also appears robust. Further, the binding of the nanocage complex to an antibody by interaction of the ZZ domain with IgG isotype antibodies is fast and effective. This can therefore be applied to a very wide range of commercially available antibodies and so can be used to effectively target a wide range of different cell types.
  • the ferritin nanocage of the invention provides a methodology for the effective delivery of compounds into cells in a phenotypic assay and the ordered assembly process is adaptable to high throughput screening scenarios. Furthermore, nanocages that are made fluorescent, either through chemical labelling, or the fusion of fluorescent proteins, can be used to monitor the uptake of individual cells. When combined with cell sorting methods the phenotypic assays could be correlated to a dose response based on the nanocage fluorescence.
  • the inventors have used phenotypic assays to demonstrate the effective delivery of the active agent Dox into cells.
  • the MTT assay measures the metabolic activity of cells via NAD(P)H dependent oxidoreductase enzymes using a tetrazolium dye substrate (MTT) that produces a purple colour on reduction. A reduced numbers of viable cells leads to a loss of activity and hence a reduced colour response.
  • MTT tetrazolium dye substrate
  • the variant ferritin polypeptides described herein maybe used to create nanocages encapsulating the test drug. In the case of the Dox loaded nanocages, two
  • concentrations of Dox ( ⁇ . ⁇ ⁇ & 0.2 ⁇ ) maybe used when forming the complexes. They may be mixed with anti-EGFR and their interaction with HT29 cells may be monitored over time prior to measuring viability using the MTT assay.
  • the nanocages that were formed with the higher loading of Dox should demonstrate a phenotypic response during the time course of the assay.
  • the data should also demonstrate a dose response to the different nanocage loading conditions used of Dox (0.1 or 2.0 ⁇ ).
  • a further phenotypic assay maybe performed using flow cytometry and a suitable dye, such as the Topro3 dye.
  • Topro3 binds to DNA and preferentially enters non-viable cells.
  • HT29 cells may be treated with Au-ZZ-GFP-hFTN (L29A L36A I81A L83A) and Dox-Au-ZZ-hFTN (L29A L36A I81A L83A) complexes pre-bound to the anti-EGFR antibody.
  • a control of Dox only may also performed along with cells only.
  • variant ferritin polypeptide of the first aspect, the fusion protein of the second aspect or the ferritin nanocage according to the third aspect i.e. which is referred to hereinafter as "agent” or “active agent"
  • agent or active agent
  • compositions having a number of different forms depending, in particular, on the manner in which the composition is to be used.
  • the composition maybe in the form of a powder, tablet, capsule, liquid etc. or any other suitable form that may be administered to a person or animal in need of treatment.
  • vehicle of medicaments according to the invention should be one which is well-tolerated by the subject to whom it is given.
  • Medicaments comprising the agents according to the invention (i.e. the ferritin nanocage) may be used in a number of ways.
  • oral administration may be required, in which case the agents may be contained within a composition that may, for example, be ingested orally in the form of a tablet, capsule or liquid.
  • Compositions comprising agents of the invention maybe administered by inhalation (e.g. intranasally).
  • Compositions may also be formulated for topical use. For instance, creams or ointments may be applied to the skin.
  • Agents according to the invention may also be incorporated within a slow- or delayed-release device.
  • Such devices may, for example, be inserted on or under the skin, and the medicament may be released over weeks or even months.
  • the device may be located at least adjacent the treatment site.
  • Such devices may be
  • agents and compositions according to the invention may be administered to a subject by injection into the blood stream or directly into a site requiring treatment. Injections maybe intravenous (bolus or infusion) or subcutaneous (bolus or infusion), or intradermal (bolus or infusion). It will be appreciated that the amount of the ferritin nanocage that is required is determined by its biological activity and bioavailability, which in turn depends on the mode of administration, the physiochemical properties of the active agent it encapsulates, if present, and whether it is being used as a monotherapy, or in a combined therapy. The frequency of administration will also be influenced by the half-lif e of the agent within the subj ect being treated.
  • Optimal dosages to be administered maybe determined by those skilled in the art, and will vary with the particular agent in use, the strength of the pharmaceutical composition, the mode of administration, and the advancement of the disease. Additional factors depending on the particular subject being treated will result in a need to adjust dosages, including subject age, weight, gender, diet, and time of administration.
  • a daily dose of between o.oiug/kg of body weight and 50omg/kg of body weight of the nanocage and/or active agent according to the invention may be used. More preferably, the daily dose is between o.oimg/kg of body weight and
  • the ferritin nanocage may be administered before, during the or after the onset of disease.
  • the nanocage maybe administered immediately after a subject has developed a disease.
  • Daily doses maybe given systemically as a single administration (e.g. a single daily injection).
  • the nanocage may require administration twice or more times during a day.
  • nanocage may be administered as two (or more depending upon the severity of the disease being treated) daily doses of between 25mg and 7000 mg (i.e. assuming a body weight of 70 kg).
  • a patient receiving treatment may take a first dose upon waking and then a second dose in the evening (if on a two dose regime) or at 3- or 4-hourly intervals thereafter.
  • a slow release device may be used to provide optimal doses of nanocage according to the invention to a patient without the need to administer repeated doses.
  • Known procedures such as those conventionally employed by the pharmaceutical industry (e.g. m ⁇ experimentation, clinical trials, etc.), may be used to form specific formulations comprising the nanocage according to the invention and precise therapeutic regimes (such as daily doses of the nanocage and/or active agent and the frequency of administration).
  • a pharmaceutical composition comprising the variant ferritin polypeptide of the first aspect, the fusion protein of the second aspect or the ferritin nanocage of the third aspect, and a pharmaceutically acceptable vehicle.
  • composition can be used in the therapeutic amelioration, prevention or treatment of any disease in a subject that is treatable, such as cancer.
  • the invention also provides, in an fourteenth aspect, a process for making the pharmaceutical composition according to the thirteenth aspect, the process comprising contacting a therapeutically effective amount of the variant ferritin polypeptide of the first aspect, the fusion protein of the second aspect or the ferritin nanocage of the first aspect, and a pharmaceutically acceptable vehicle.
  • a "subject” maybe a vertebrate, mammal, or domestic animal.
  • agents, compositions and medicaments according to the invention may be used to treat any mammal, for example livestock (e.g. a horse), pets, or maybe used in other mammals.
  • the subject is a human being.
  • a “therapeutically effective amount” of agent is any amount which, when it is not
  • administered to a subject is the amount of drug that is needed to treat the target disease, or produce the desired effect, e.g. result in tumour killing.
  • the therapeutically effective amount of nanocage and/or active agent used maybe from about o.oi mg to about 8oo mg, and preferably from about o.oi mg to about 500 mg.
  • a "pharmaceutically acceptable vehicle” as referred to herein, is any known
  • the pharmaceutically acceptable vehicle maybe a solid, and the composition may be in the form of a powder or tablet.
  • a solid pharmaceutically acceptable vehicle may include one or more substances which may also act as flavouring agents, lubricants, solubilisers, suspending agents, dyes, fillers, glidants, compression aids, inert binders, sweeteners, preservatives, dyes, coatings, or tablet- disintegrating agents.
  • the vehicle may also be an encapsulating material.
  • the vehicle is a finely divided solid that is in admixture with the finely divided active agents according to the invention.
  • the nanocage maybe mixed with a vehicle having the necessary compression properties in suitable proportions and compacted in the shape and size desired.
  • the powders and tablets preferably contain up to 99% of the active agents.
  • Suitable solid vehicles include, for example calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose,
  • the pharmaceutical vehicle may be a gel and the composition may be in the form of a cream or the like.
  • the pharmaceutical vehicle maybe a liquid, and the pharmaceutical composition is in the form of a solution.
  • Liquid vehicles are used in preparing solutions, suspensions, emulsions, syrups, elixirs and pressurized compositions.
  • the nanocage maybe dissolved or suspended in a pharmaceutically acceptable liquid vehicle such as water, an organic solvent, a mixture of both or pharmaceutically acceptable oils or fats.
  • the liquid vehicle can contain other suitable pharmaceutical additives such as solubilisers, emulsifiers, buffers, preservatives, sweeteners, flavouring agents, suspending agents, thickening agents, colours, viscosity regulators, stabilizers or osmo- regulators.
  • suitable pharmaceutical additives such as solubilisers, emulsifiers, buffers, preservatives, sweeteners, flavouring agents, suspending agents, thickening agents, colours, viscosity regulators, stabilizers or osmo- regulators.
  • suitable examples of liquid vehicles for oral and parenteral administration include water (partially containing additives as above, e.g. cellulose derivatives, preferably sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g. glycols) and their derivatives, and oils (e.g.
  • the vehicle can also be an oily ester such as ethyl oleate and isopropyl myristate.
  • Sterile liquid vehicles are useful in sterile liquid form compositions for parenteral administration.
  • the liquid vehicle for pressurized compositions can be a halogenated hydrocarbon or other pharmaceutically acceptable propellant.
  • Liquid pharmaceutical compositions which are sterile solutions or suspensions, can be utilized by, for example, intramuscular, intrathecal, epidural, intraperitoneal, intravenous and particularly subcutaneous injection.
  • the nanocage may be prepared as a sterile solid composition that maybe dissolved or suspended at the time of administration using sterile water, saline, or other appropriate sterile injectable medium.
  • compositions of the invention may be administered orally in the form of a sterile solution or suspension containing other solutes or suspending agents (for example, enough saline or glucose to make the solution isotonic), bile salts, acacia, gelatin, sorbitan monoleate, polysorbate 8o (oleate esters of sorbitol and its anhydrides copolymerized with ethylene oxide) and the like.
  • solutes or suspending agents for example, enough saline or glucose to make the solution isotonic
  • bile salts for example, enough saline or glucose to make the solution isotonic
  • bile salts for example, enough saline or glucose to make the solution isotonic
  • acacia gelatin
  • sorbitan monoleate sorbitan monoleate
  • polysorbate 8o oleate esters of sorbitol and its anhydrides copolymerized with ethylene oxide
  • the nanocage according to the invention
  • Forms useful for parenteral administration include sterile solutions, emulsions, and suspensions.
  • the skilled technician will appreciate that in order to calculate the percentage identity between two DNA/polynucleotide/nucleic acid sequences, an alignment of the two sequences must first be prepared, followed by calculation of the sequence identity value.
  • the percentage identity for two sequences may take different values depending on:- (i) the method used to align the sequences, for example, QustalW, BLAST, FASTA, Smith- Waterman (implemented in different programs), or structural alignment from 3D comparison; and (ii) the parameters used by the alignment method, for example, local vs global alignment, the pair-score matrix used (e.g. BLOSUM62, PAM250, Gonnet etc.), and gap-penalty, e.g. functional form and constants.
  • percentage identity between the two sequences. For example, one may divide the number of identities by: (i) the length of shortest sequence; (ii) the length of alignment; (Hi) the mean length of sequence; (iv) the number of non-gap positions; or (iv) the number of equivalenced positions excluding overhangs. Furthermore, it will be appreciated that percentage identity is also strongly length dependent Therefore, the shorter a pair of sequences is, the higher the sequence identity one may expect to occur by chance.
  • DNA/polynucleotide/nucleic acid sequences is then calculated from such an alignment as (N/T)*ioo, where N is the number of positions at which the sequences share an identical residue, and T is the total number of positions compared including gaps but excluding overhangs.
  • nucleotide/nucleic acid sequence will be encoded by a sequence which hybridizes to the sequences shown in any one of SEQ ID Nos. l to io, or their complements under stringent conditions.
  • stringent conditions we mean the nucleotide hybridises to filter-bound DNA or R A in 3x sodium
  • SSC chloride/sodium citrate
  • nucleic acid sequence could be varied or changed without substantially affecting the sequence of the protein encoded thereby, to provide a functional variant thereof.
  • Suitable nucleotide variants are those having a sequence altered by the substitution of different codons that encode the same amino acid within the sequence, thus producing a silent change.
  • Other suitable variants are those having homologous nucleotide sequences but comprising all, or portions of, sequence, which are altered by the substitution of different codons that encode an amino acid with a side chain of similar biophysical properties to the amino acid it substitutes, to produce a conservative change.
  • small non-polar, hydrophobic amino acids include glycine, alanine, leucine, isoleucine, valine, proline, and methionine.
  • Large non-polar, hydrophobic amino acids include phenylalanine, tryptophan and tyrosine.
  • the polar neutral amino acids include serine, threonine, cysteine, asparagine and glutamine.
  • the positively charged (basic) amino acids include lysine, arginine and histidine.
  • the negatively charged (acidic) amino acids include aspartic acid and glutamic acid. It will therefore be appreciated which amino acids may be replaced with an amino acid having similar biophysical properties, and the skilled technician will know the nucleotide sequences encoding these amino acids.
  • the black arrow that intersects the x-axis at 5.79 ml shows the elution point of commercial 24- meric horse spleen ferritin.
  • the dark blue and red lines correspond to the absorbance readings at 280 nm and 420 nm respectively.
  • the light blue and red shading corresponds to ⁇ 1 standard deviation of the mean absorbance readings at 280 nm (protein) and 420 nm (heme), respectively.
  • Each data set is composed of three biological repeats;
  • Figure 2 shows the results of size exclusion chromatography of Bfr with Au
  • Figure 4 shows dimeric interfaces in light chain ferritin QFTN) and heavy chain ferritin (hFTN).
  • A.1FTN dimer (PDB ID:2FG8 (asymmetric unit) [156]).
  • B. hFTN dimer (PDB ID: 3AJO (biological assembly 1) [158]). For each dimer, one subunit is shown in orange and the other is shown in blue.
  • C.1FTN dimer highlighting the conserved hydrophobic residues in the dimer interface and the list of mutations.
  • D. hFTN dimer highlighting the conserved hydrophobic residues in the dimer interface.
  • Figure 8 shows behaviour of hFTN.
  • Figure 9 shows reassembly of mutant hFTN.
  • the 24-mer elutes at approximately 5.3 ml and the monomer elutes at approximately 6.8 ml.
  • the wt hFTN is seen to elute primarily as 24- mer (panel A).
  • the AuNP co -elutes with the FTN 24-mer.
  • the dark green line corresponds to the absorbance readings at 497 nm (GFP absorbance) and the dark blue line absorbance at 530 nm (AuNP absorbance).
  • the shading in both instances corresponds to ⁇ 1 standard deviation of the mean absorbance readings.
  • Each dataset is comprised of three biological repeats;
  • Figure 10 shows the results of TEM analysis of hFTN with AuNP.
  • TEM analysis of hFTN with AuNP TEM analysis of hFTN with AuNP.
  • A wt ZZ-GFP-hFTN with AuNP, blue arrows indicate clusters with AuNP, red arrows indicate isolated nanocages;
  • B mutant ZZ-GFP-hFTN (L29A L36A I81A L83A) with AuNP, blue arrows indicate nanocages with encapsulated AuNP, red arrows indicate isolated nanocage fragments, yellow arrows indicate empty nanocages;
  • C mutant ZZ-GFP-hFTN (L29A L36A I81A L83A) without AuNP
  • D wt ZZ-GFP- hFTN without AuNP, red arrows indicate nanocages;
  • Figure 11 shows the binding of Doxorubicin to gold nanoparticles.
  • the binding of doxorubicin (Dox) to 5 nm gold nanoparticles was monitored from the fluorescence signal of the Dox.
  • a titration of Dox concentration was measured in PBS either in the presence or absence of 5 nm Au nanoparticles.
  • Fluorescence was measured in a BMG Clariostar plate reader (ex: 482-16; emm: 580-30) and intensity plotted after subtraction of background. Binding of the Dox to the Au causes a significant quenching of the Dox fluorescence;
  • Figure 12 shows the interaction of propidium iodide with Au nanoparticles.
  • the binding of propidium iodide (PI) to 5 nm gold nanoparticles was monitored from the fluorescence signal of the PI.
  • a titration of PI concentration was measured in PBS either in the presence or absence of 5 nm Au nanoparticles.
  • Fluorescence was measured in a Fluoromax-4 (ex: 493 nm; emm: 550-750) and emission scans are plotted after subtraction of background. Binding of the PI to the Au causes complete ablation of the PI fluorescence;
  • Figure 13 shows Dox fluorescence in purified nanocage-Au-Dox complexes.
  • Figure 14 is mass spectrometry analysis of drug encapsulation.
  • Complexes containing hFTN (L29A L36A I81A L83A), Au nanoparticle and Dox were formed by adding the mutant ferritin protein (0.1 ⁇ ) to different concentrations of Dox (0.1 ⁇ to 10.0 ⁇ ), Au nanoparticle preparations stabilised with either citrate or PBS (phosphate buffered saline) were used to evaluate if this affected the binding of the drug to the gold.
  • the nanocages formed were purified by HPLC and analysed by LC-MS (Agilent 6550), data were quantified using a 20 ppm window for Dox and PI based on a calibrated standard;
  • Figure 15 shows antibody directed cell binding of GFP nanocage.
  • Purified wt ZZ-GFP- hFTN (20 ⁇ g) was mixed with either anti-NKi.i antibody (1 ⁇ g) or anti-EGFR antibody (1 ⁇ g) in 210 ⁇ l of PBS.
  • 50 ⁇ l of the nanocage-antibody was mixed with 1 x 10 6 cells of either HT29 or MNK1.1 in 100 ⁇ l.
  • Cells were analysed an a BD Fortessa using the FITC channel (ex 488 nm; emm 530-30 nm) to observe GFP fluorescence.
  • Figure 17 shows delivery of Dox to cells by encapsulated nanocage. Confocal microscopy showing a z-slice. Purified Dox-Au-ZZ-hFTN (L29A L36A I81A L83A) (100 ⁇ l of 30 nM) was mixed with anti-EGFR antibody (1 ⁇ g) in 210 ⁇ l of PBS. HT-29 cells were seeded on chamber slides (ibidi) in DMEM medium with 10% FBS overnight for cell attachment. Cells were then treated with the nanocage-antibody complex (100 ⁇ l) at 37 °C for different times (panels a-c, 2 h, d-f, 24 h).
  • Figure 19 shows purified Dox/PI-Au-ZZ-hF (L29A L36A I81A L83A) (100 ⁇ l of ⁇ 30 nM) was mixed with anti-EGFR antibody (1 ⁇ g) in 210 ⁇ l of PBS.
  • HT-29 cells were grown in DMEM medium with 10% FBS overnight. Cells were then treated with the nanocage-antibody complex (100 ⁇ l) at 37 °C for different 48 h and 72 h. After incubation, the cells were washed 3x with cold PBS. Re-suspended cells were analysed by LC-MS (Agilent 6550), data were quantified using a 20 ppm window for Dox and PI based on a calibrated standard;
  • Figure 20 shows phenotypic assays of drug delivery, a) MTT assay.
  • Purified Dox-Au- ZZ-hFT (L29A L36A I81A L83A) (100 ⁇ l of 30 nM), prepared by loading with either 0.1 ⁇ M or 2.0 ⁇ M DOX, was mixed with anti-EGFR antibody (1 ⁇ g) in 210 ⁇ l of PBS. Cells were cultured on a three 96 well plate (5000 cells/well) Then, cells were incubated with the prepared nanocage-antibody complexes.
  • Figure 21 shows a phenotypic cell killing assay using Vybrant staining and flow cytometry for nanocage delivered Paclitaxel (Pac).
  • Purified Pac-Au-ZZ-hFTN (L29A L36A I81A L83A) (100 ⁇ l of 30 nM), was prepared by loading with 5.0 ⁇ M Pac and unincorporated drug removed by Zorbax spin column; this was mixed with anti-EGFR antibody (1 ⁇ g) in PBS and exposed to 5x10 5 live cells.
  • A shows the degree of dead cells observed after 24I1 and 48I1 for the drug loaded nanocage in the absence of antibody, in the presence of antibody and for 5 ⁇ free drug.
  • B shows the flow cytometry dot plots for cells only, cells with hFtn only, free drug only and Pac loaded nanocage with antibody; the upper left quadrant shows dead cells and the lower left live cells;
  • Figure 22 shows a phenotypic cell killing assay using Vybrant staining and flow cytometry for nanocage delivered Actinomycin-D (Act-D).
  • Purified Act-D-Au-ZZ-hFTN (L29A L36A I81A L83A) (100 ⁇ l of 30 nM), was prepared by loading with 5.0 ⁇ M Act-D and unincorporated drug removed by Zorbax spin column; this was mixed with anti- EGFR antibody (1 ⁇ g) in PBS and exposed to 5x10s live cells.
  • A shows the degree of dead cells observed after 24I1 and 48I1 for the drug loaded nanocage in the absence of antibody, in the presence of antibody and for 5 ⁇ free drug.
  • B shows the flow cytometry dot plots for cells only, cells with hFtn only, free drug only and Act-D loaded nanocage with antibody; the upper left quadrant shows dead cells and the lower left live cells; and Figure 23 shows mass spectrometry results performed to determine quantitation of Act-D as encapsulated within the hFtn nanocage.
  • a calibration curve was performed for the monomer His-ZZ-hFTN(L2gA L36A I81A L83A) based on the QNYHQDSEAAINR peptide.
  • B A calibration curve was performed for Actinomycin-D bound to Au nanoparticles.
  • thermostable ferritin from Archaeoglobus fidgidus ⁇ A.fu is stable in a dimeric form at low salt and reversibly forms nanocage structures on transition to high salt 15, 16 .
  • it could interact with a gold nanoparticle to form a ferritin-encapsulated gold nanoparticle.
  • Other efforts to encapsulate either drugs or metal cores into ferritin rely on the fact that it dissociates into its constituent dimers at low pH and can reform the nanocage on transition back to neutral pH3. v, 18. However, this pH change is also partially destructive and it impacts the integrity of the reformed nanocage 18 .
  • a plasmid encoding the recombinant protein of interest was transformed into E. coli BL21-DE3. Single colonies were suspended in 8 x 5 mL LB media containing
  • Pellet cells were thawed on ice in lysis buffer (1 x PBS, 50 mM imidazole, 100 mM NaCl, pH 7.2) containing 1 protease inhibitor cocktail tablet (Roche). Resuspended cells were sonicated for 2 x 10 mins (amplitude 40 %, pulse 2 seconds on 2 seconds off) and then centrifuged (15000 rpm, 4 °C, 40 min.). Initial purification conducted with
  • His-tag immobilized metal ion affinity chromatography
  • His-tag beads chelating sepharose fast flow, GE healthcare
  • Eluted protein was dialysed overnight (100 mM NaQ, 1 x PBS, pH 7.2). Protein was concentrated to 1-2 mL using Amicon ultra- 15 centrifugal filter unit (3000 rpm, 4 °C, ⁇ 30 mins). Further purification was conducted using size exclusion chromatography. GE Akta FPLC system combined using a
  • SEC size exclusion chromatography
  • HPLC high performance liquid chromatography
  • the concentration of protein samples was calculated using absorbance spectroscopy with an extinction coefficient of 15,930 cm 1 M 1 at 280 nm for human light chain ferritin and 18,910 cm 1 M 1 at 280 nm for human heavy chain ferritin. Extinction coefficients for other fusion proteins, extinction coefficients were calculated using the ExPASy ProtParam tool. The ratio of Bfr subunits to heme molecules was calculated using an extinction coefficient for heme of 137,000 cm 1 M 1 at 417 nm.
  • the purified ferritin protein was mixed with 5 nm gold nanoparticles (Sigma Aldrich). Stoichiometry was estimated from protein concentration and stated number of gold particles per unit volume, calculated to give 24 protein monomers per gold
  • nanoparticles at the concentration indicated at room temperature ferritin was added between 1 min and 30 min after.
  • Gold nanoparticles and protein were co-incubated for 12 hours at 4 °C. If needed concentrated to 1-2 mL (3000 rpm, 4 °C) and then purified using HPLC size exclusion chromatography, as above. Fractions containing nanocage were combined and concentrated to 1-2 mL (3000 rpm, 4 °C, ⁇ i hour). When used for storage mixed equally (by volume) with 80 % glycerol.
  • Concentrations of gold nanoparticle encapsulated ferritin nanocages were calculated based on the sum of the extinction coefficient at 280 nm of 5 nm gold nanoparticles (1.66 xio 7 M ⁇ cnr 1 ) and the extinction coefficient at 280 nm for the relevant protein components also present.
  • Protein samples were mounted on carbon coated copper grids.
  • the grids were prepared in advance using glow discharge. This technique increases the hydrophilicy of the grid allowing the protein sample to adhere to the carbon coating. After the protein sample had been loaded onto the grid, a negative stain was applied (uranyl acetate) to provide contrast.
  • Fluorescence measurements were performed either on a Jobin Yvon Fluoromax 4 with a 400 ⁇ l cuvette using excitation and emission wavelengths as stated and slit widths of 5 nm. Alternatively a BMG Clariostar plate reader was used with filters or
  • LCMS Chromatographic MS
  • Agilent 6550 LC separation was achieved using a 1290 Infinity system (Agilent, Santa Clara, CA) and a Vydac 214MS C4 column, 2.1x150mm and sum particle size, (Grace, Columbia, MD) at a temperature of 35°C with a buffer flow rate of o.2ml/min. with a denaturing mobile phase: buffer A was o.1% formic acid in water and buffer B was o.1% formic acid in acetonitrile. Elution of components was achieved using a linear gradient from 3% to 40% buffer B over 18.5 min.
  • On-line mass spectra were accumulated on a 6550 quadrupole time-of-flight instrument with a dual electrospray Jet Stream source (Agilent). Mass spectra were acquired of the m/z range of 100-1700 at a rate of 0.6 spectra per second. Targeted MS/MS were acquired over the range of 100-1700 Da with a 1.3 Da precursor isolation window and a collision energy of lseV.
  • Nanocage was incubated in 8M Urea in 50mM Tris-HCl (pH 8) with 4mM DTT and heated at 95°C for 20 minutes. After denaturation the reaction mixture was cooled and 50mM NH 4 HC0 3 was added such that the urea concentration is below lM. Modified Trypsin was then added to a final proteaseiprotein ratio of 1:100 and incubated overnight at 37°C for complete digestion. Human Ferritin mutant monomer samples did not require urea denaturation and were only digested with Trypsin.
  • Standard solutions of varying concentrations (o, 0.05, 0.1, 0.2, 0.5, 1, 2 ⁇ M) were prepared for the drug in buffer, drug on gold nanoparticles and human Ferritin mutant monomer.
  • the targeted LC-MS/MS method was applied using an Agilent 1290 LC system coupled to an Agilent 6550 quadrupole - time-of-flight (Q-ToF) mass spectrometer with electrospray ionization (Agilent, Santa Clara, CA).
  • the LC column used was an Agilent Zorbax Extend C-18, 2.1 x 50mm and i.8um particle size.
  • the LC buffers were 0.1% formic acid in water and 0.1% formic acid in acetonitrile (v/v).
  • two diagnostic tryptic peptides for the protein to be measured were selected for the targeted LC-MS/MS method.
  • Quantitation was based on the LC retention times of standards and the area of accurately measured diagnostic precursor or fragment ions.
  • the protonated molecules of each peptide, [M+2H] 2+ were targeted and subjected to collision induced dissociation, with product ions accumulated throughout the targeted period.
  • Concentrations were calculated using the integrated area of the peak corresponding to the elution of the molecule or peptide of interest at the retention time of the standards.
  • Flow cytometry was performed on a BD Fortessa using the FITC channel to observe GFP (ex 488 nm; emm 530-30 nm; ToPro-3 was imaged in red channel (640 nm ex; 670/14 emission). Data was analysed using Flow-Jo software. Cell preparation for LCMS analysis
  • Cells were lysed using a bead beading process. Cells were pelleted at 7k rcf for 10 min. and dissolved in 100 ⁇ l methanol and vortexed until homogenous.50 ⁇ l of acid washed glass beads (Sigma) were added. Cells were then vortexed for 30s and kept on ice for 30s four times before centrifugation at 14 krpm at 4°C for 15 min. Supernatant was then taken for LCMS analysis as above.
  • Cells were washed twice with PBS and fixed with 4 % formaldehyde for 10 minutes and then washed 3 x with PBS. Cells were then permeabilised with 0.1 % TX-100/PBS for 15-20 minutes and wash 3 x. Cells were then blocked with 5 % normal goat serum/PBS or 1 % BSA/PBS for 45 minutes (no washing required). The primary antibody was diluted in blocking solution and applied for 2 h (or overnight at 4 °C). Wash 4 x thoroughly to remove unbound primary antibody. Cells were then incubatee with the secondary antibody for 1 h, diluted in blocking solution or wash buffer.
  • the secondary antibody was then aspirated and, if required, incubated with DAPI [1 ug/mL] in PBS for 10 minutes and washed 4 x. Coverslip was then dipped into H 2 0 to remove residual salts of the wash buffer. A drop of mounting medium was added and the slide sealed. Antibodies used were as stated in Figure legends.
  • Dox-Au-ZZ-hFTN (L29A L36A I81A L83A) (100 ⁇ l of 31 nM) was prepared by loading with either 0.1 ⁇ M or 2.0 ⁇ M DOX, was mixed with anti-EGFR antibody (1 ⁇ g) in 210 ⁇ l of PBS. Cells were cultured on a three 96 well plate (5000 cells/well) Then, cells were incubated with nanocage constructs to be tested.
  • Dox-Au-ZZ-hFTN (L29A L36A I81A L83A) (100 ⁇ l of 31 nM) was prepared by loading with 2.0 ⁇ M DOX, was mixed with anti-EGFR antibody (1 ⁇ g) in 210 ⁇ l of PBS. Cells were cultured on a three 96 well plate (5000 cells/well) Then, cells were incubated with nanocage constructs to be tested. Prior to assay, cells were mixed with ToPro-3 staining solution (1 ⁇ M) and incubated for 30 min, washed with PBS and analysed on a BD Fortessa (640 nm ex; 670/14 emission), data was analysed using FlowJo. Phenotvpic cell death assays using Vvbrant cell staining assay
  • HT-29 cells were trypsinised and cell viability was assessed using the Trypan blue exclusion assay - count cells treated with Trypan blue dye using a haemocytometer, and determine the volume of cell suspension that contains 5X10 5 live cells.
  • Live cells (5X105) seeded into a 12-well plate with a final volume of 500 ⁇ L. This final 500 ⁇ L volume will consist of 5X105 cells + medium + drug-Au-ZZ-hFTN + anti-EGFR antibody (0.5 - 1 ⁇ g).
  • the plate was incubated in a tissue culture incubator set at 37°C, 5% CO2, 95% humidity for 24 h or 48 h before the experiment was stopped.
  • Uptake of drug-Au-ZZ-hFTN by cells was stopped by removing the 500 ⁇ L solution containing test or control compounds, trypsinising cells and pelleting cells in preparation for cytotoxicity assays, e.g. Vybrant cell apoptosis assay. Prior to staining the cells were centrifuged (3000 rpm for 2 min) and then washed in cold PBS before a second centrifugation step at 3000 rpm for 2 min in a microcentrifuge. The supernatant was removed and discarded before the cell pellet was resuspended in 1 mL ice cold sterile lX PBS containing YOPRO and PI stain.
  • cytotoxicity assays e.g. Vybrant cell apoptosis assay.
  • the stained cells were analysed by flow cytometry, using 488 nm excitation with green fluorescence emission for YOPRO R-i (i.e., 530/30 bandpass) and red fluorescence emission for propidium iodide (i.e., 610/20 bandpass), gating on cells to exclude debris.
  • Single-color stained cells were used to perform standard compensation. The stained cell population will separate into three groups: live cells show a low level of green fluorescence, apoptotic cells show an incrementally higher level of green fluorescence, and dead cells show both red and green fluorescence.
  • HFn nanocage The cellular uptake and distribution of HFn nanocage were studied by confocal microscope (Zeiss LSM 510). Briefly, HT-29 cells were seeded on chamber slides (ibidi) in DMEM medium with 10% FBS overnight for cell attachment. Cells were then treated with HFn at 37 °C for different times. After the incubation, the cells were washed with cold PBS, fixed in 4% cold Paraformaldehyde, and permeabilized with 0.1% Triton X- 100. To visualize lysosomes, the cells were further incubated with an anti-Lampi (1:100; Biolegend) for 1 h after blocking by 1% BSA .
  • ferritin from different biological sources bacterioferritin (Bfr) was isolated from E. coli and contains 24 subunits and 12 heme groups that bind between the dimeric protein interface.
  • Human ferritin (FTN) can be composed of the light chain ferritin subunit (1FTN) or heavy chain ferritin subunit (hFTN), or a combination of both. By expressing either 1FTN or hFTN in E. coli it is possible to create ferritin nanocages that consist of only a single protein monomer.
  • Protein samples were mounted on carbon coated copper grids.
  • the grids were prepared in advance using glow discharge. This technique increases the hydrophilicity of the grid allowing the protein sample to adhere to the carbon coating.
  • a negative stain was applied (uranyl acetate) to provide contrast. After staining, the samples were imaged using transmission electron microscopy (TEM).
  • TEM transmission electron microscopy
  • the bfr gene was amplified from the E. coli genome and cloned into an expressing construct. Two variants of the gene were generated, one (SEQ ID No.5) included an N-terminal His tag for purification, and the second (SEQ ID No. 9) contained a C-terminal gold binding peptide (AuBP). Metal binding peptides have been shown to provide a mechanism for coordinating the binding of proteins to metallic surfaces 1 ' and it had been shown that the addition of the Au binding peptide could facilitate the
  • hFTN As the destabilisation of hFTN worked well, a domain was added to its N-terminus to facilitate its subsequent binding to antibodies.
  • Z-domain was chosen. This is a derivative of Staphylococcus protein A, and is an engineered version of the IgG binding domain of protein A with greater stability and a higher binding affinity for the Fc antibody domain (Nilsson 1987, ref 21).
  • the Z domain was coded as a repeat so that two tandem domains would be present (ZZ).
  • SEC analysis of hFTN with an N- terminal ZZ and GFP demonstrates that the full length protein is still purified as a nanocage, while the mutated hFTN purifies as a monomer (see Figure 7).
  • Example 2 the inventors wanted to demonstrate if they could reassemble the nanocage in an ordered manner around a metallic nanoparticle (e.g. gold), as they had done previously with Bfr (see Figure 3 - Example 1).
  • the ZZ-GFP-FTN fusions for both wild type hFTN and mutant hFTN (L29A L36A I81A L83A) were incubated with
  • AuNP gold nanoparticle
  • SEC size exclusion chromatography
  • the ZZ- GFP-hFTN (L29A L36A 18 lA L83A) mutant does not form nanocages in the absence of AuNP (see Figure 10C), but in the presence of AuNP there is a high proportion of nanocage structures where the AuNP is clearly encapsulated within the central space of the ferritin nanocage (see Figure 10B).
  • Example 3 the inventors have demonstrated the ordered assembly of the ferritin nanocages around a gold nanoparticle. They have also used this programmed ordered assembly to enable the direct encapsulation of drugs inside the nanocages.
  • Gold nanoparticles have been considered as stand-alone vectors for drug delivery through the formation of covalent drug-Au conjugates 20 .
  • Dox anti-cancer drug doxorubicin
  • Quenching of the fluorescence in the presence of Au nanoparticles demonstrates an interaction between the Dox and the Au (see Figure 11). In addition, they demonstrated an interaction between propidium iodide (PI) and Au nanoparticles, and in this instance a complete ablation of fluorescence was observed (see Figure 12).
  • PI propidium iodide
  • the mutant hFTN (L29A L36A I81A L83A) protein was added to the Au nanoparticles in the presence of different concentrations of Dox or PI.
  • the nanocages that were formed around the Au nanoparticle were then purified by HPLC (as in Figure 9).
  • the purified Dox-Au- nanocage complex was then evaluated for Dox by measurement of Dox fluorescence. The clear presence of Dox fluorescence indicated that Dox was present in the purified nanocage complexes (see Figure 13). Encapsulation of PI by fluorescence could not be monitored due to its complete quenching on binding.
  • Ferritin fusions containing an N -terminal ZZ domain should be able to bind to IgG isotype antibodies since the Z-domain is a synthetic derivative of an IgG binding domain from Staphylococcus aureus protein A.
  • the inventors evaluated the specificity with which they can direct the targeting of the ferritin nanocage to specific cell types by direct antibody interactions. To establish a fluorescent basis for determining cell binding they used the GFP labelled wt ZZ-GFP-hFTN.
  • MNK1.1 mouse natural killer cells
  • HT29 colonal cancer
  • Nanocages with GFP were created to monitor the delivery and fate of the nanocage in cells, while ferritin without GFP was used to create nanocages with Au- drug encapsulated so that the fate of the drug could be monitored by fluorescence.
  • Au- ZZ-GFP-hFTN (L29A L36A I81A L83A) and Drug-Au-ZZ-hFTN (L29A L36A I81A L83A) complexes were formed as before and purified by HPLC. They were then mixed with anti-EGFR as before and their interaction with HT29 cells was monitored over time.
  • the GFP-labelled nanocages were clearly seen to bind to the cells and after 2 h punctate distributions of nanocages could be observed both on the surface and inside the cells (F g. 16). Cells were also stained with lampi, a late lysosomal marker. The internalised GFP signal after 2h can clearly be seen to be punctate but not associate with lysosomes, consistent with early stage endocytosis into endosomes (see Figures 16a and 16b). After 24I1, the picture clearly changed, with GFP being dispersed throughout the cell cytoplasm and partly associated with lysosomal signal, consistent with it being broken down and dispersed by the pH drop associated with lysosomes (see Figures i6d and i6e).
  • the inventors have used phenotypic assays to demonstrate the effective delivery of Dox into cells.
  • the MTT assay measures the metabolic activity of cells via NAD(P)H dependent oxidoreductase enzymes using a tetrazolium dye substrate (MTT) that produces a purple colour on reduction. A reduced numbers of viable cells leads to a loss of activity and hence a reduced colour response.
  • Au-ZZ-GFP-hFTN (L29A L36A I81A L83A) and Dox-Au-ZZ-hFT (L29A L36A 18 lA L83A) complexes were formed as before and purified by HPLC. In the case of the Dox loaded nanocages, two
  • concentrations of Dox (0.1 ⁇ & 0.2 ⁇ ) were used when forming the complexes. They were then mixed with anti-EGFR as before and their interaction with HT29 cells was monitored over time prior to measuring viability using the MTT assay.
  • the nanocages that were formed with the higher loading of Dox clearly demonstrated a phenotypic response during the time course of the assay (Fig. 20a).
  • the data also demonstrate a dose response to the different nanocage loading conditions used of Dox (0.1 or 2.0 ⁇ ).
  • a further phenotypic assay was performed using flow cytometry and the Topro3 dye. Topro3 binds to DNA and preferentially enters non-viable cells.
  • HT29 cells were treated with Au-ZZ-GFP-hFTN (L29A L36A 18 lA L83A) and Dox-Au-ZZ-hFTN (L29A L36A I81A L83A) complexes pre-bound to the anti-EGFR antibody; a control of Dox only was also performed along with cells only (Fig. 20b).
  • the drug loaded nanocage demonstrates a clear difference in viability at 24I1.
  • the difference with the control cells becomes less pronounced at longer time points, and this may be due to uptake being triggered by the presence of the anti-EGFR antibody. It is also known that at longer time points this dye becomes less specific as a viability signal, although the cell only control has a low response even after 72I1.
  • Example 8 Using the nanocage in a phenotypic screening platform
  • the inventors have demonstrated the ability to use the ferritin nanocage as a platform technology for the delivery of small molecule drugs into cells. Because the technology provides a defined process for the encapsulation and assembly of the nanocage complex, it can be envisioned as a generic method for the delivery of compounds into cells.
  • the binding of small molecule compounds to the Au nanoparticle will work for a wide variety of ionic, electrostatic and hydrophobic interactions.
  • the assembly of the mutant nanocage around the drug-bound nanoparticle also appears robust.
  • the binding of the nanocage complex to an antibody by interaction of the ZZ domain with IgG isotype antibodies is fast and effective. This can therefore be applied to a very wide range of commercially available antibodies and so can be used to effectively target a wide range of different cell types.
  • the ferritin nanocage described herein provides a methodology for the effective delivery of compounds into cells in a phenotypic assay and the ordered assembly process is adaptable to high throughput screening scenarios. Furthermore, nanocages that are made fluorescent, either through chemical labelling, or the fusion of fluorescent proteins, can be used to monitor the uptake of individual cells. When combined with cell sorting methods the phenotypic assays could be correlated to a dose response based on the nanocage fluorescence.
  • ferritin nanocages to specific cell types via the binding of antibodies creates possibilities for the diagnosis and treatment of disease. Because the nanocages can be made fluorescent, they can be used in imaging methods to identify specific cell types displaying known epitope disease markers. This creates possibilities for their use in the diagnosis of cancer types in imaging accessible locations. Examples of this are cancers accessible via Gl-tract, such as oesophageal, stomach, colorectal, liver, pancreatic, gall bladder. In addition, cancers near to the surface of the body would be accessible for diagnosis including skin cancer and neck and throat cancers.
  • the ability to encapsulate drugs into the nanocage also provides the possibility of combined diagnostic and therapy (theranostic) approaches.
  • the drug encapsulated complex contains an Au nanoparticle, a mechanism for the activated release of drugs is also possible.
  • Au nanoparticles absorb light due to their plasmonic effect and laser irradiation is proven to cause localised heating of the nanoparticle proportional to the intensity of the incident laser irradiation (Honda et al).
  • laser induced heating may therefore be used to activate the release of the encapsulated drug, since localised heating will lead to the thermal disassembly of the nanocage complex.
  • This type of approach can make use of current endoscope technology that can both locally deliver compounds, image and treat using laser light sources. The inventors therefore consider that this type of nanocage device would fit with current therapeutic practices and approaches.
  • the principle of laser-induced drug release can be demonstrated by examining the fluorescence polarisation of a fluorescently bound molecule within the nanocage, such as Dox.
  • Anisotropy provides an intensity independent measure of the degree of polarisation within a sample. Briefly, when a fluorescent molecule absorbs plane polarised light, it will be emitted in the same plane as the excitation source. However, during the fluorescence lifetime, between absorption and emission, the molecule may rotate. This means that the emitted light will be relative to the new orientation of the molecule. By measuring the emitted light in both vertical and horizontal planes, it is possible to determine the degree of polarisation (anisotropy).
  • Paclitaxel is a natural product, first isolated from the Pacific yew tree. It is commonly used to treat many types of cancer and is known to have many side effects. It prevents cell division by targeting mitotic spindle assembly.
  • An albumin bound formulation (abraxane) has, to a degree, enhanced the efficacy of the drug in cancer treatment, and alleviated some of the toxicity issues associated with the solvent previously used for administration.
  • Abraxane demonstrates, in principle, the advantages that can be obtained for appropriate drug delivery, but it still has significant toxicity issues. The inventors have performed experiments to demonstrate the encapsulation and delivery of Pac by the ferritin nanocage of the invention to a colon tumour cancer cell line - HT-29.
  • Pac (5 ⁇ ) was encapsulated to create Drug-Au-ZZ-hFtn(L29A L36A L81A L83A) nanocages as described above. Excess free drug was removed using a Zorbax spin column prior to addition to cells.
  • Anti-EGFR antibody (0.5 ⁇ g) was added to the Pac-Au-ZZ-hFtn(L29A L36A L81A L83A) and the antibody bound cage added to cells (30 nM).
  • the unloaded Au-ZZ-hFtn(L29A L36A L81A L83A) delivery vehicle was added to HT29 cells as a control to determine cytotoxic effects of hFtn that only contained gold nanoparticles.
  • Free Pac was added to cells at high concentration (5 ⁇ M) as a drug only control.
  • the phenotypic effect of the delivery of drugs into cells was assessed via Vybrant fluorescent staining using flow cytometry to measure percentages of dead, apoptotic and live cells.
  • Actinomycin-D (Act-D) consists of two cyclic peptides linked via a phenoxazone ring. It is an antibiotic that is also used as a chemotherapy medication to treat a number of types of cancer and is on the WHOs list of essential medicines. It has significant side effects.
  • the inventors performed experiments to discover if a cyclic peptide of the size and complexity of Act-D could be encapsulated and delivered to cells by the ferritin nanocage of the invention to a colon tumour cancer cell line - HT-29.
  • Act-D (5 ⁇ M) was encapsulated to create Drug-Au-ZZ-hFtn(L29A L36A L81A L83A) nanocages as before. Excess free drug was removed using a Zorbax spin column prior to addition to cells.
  • Anti-EGFR antibody 0.5 ⁇ g was added to the Act-D-Au-ZZ-hFtn(L29A L36A L81A L83A) and the antibody bound cage added to cells (30 nM).
  • the unloaded Au-ZZ-hFtn(L29A L36A L81A L83A) delivery vehicle was added to HT29 cells as a control to determine cytotoxic effects of hFtn that only contained gold nanoparticles.
  • Free Act-D was added to cells at high concentration (5 ⁇ M) as a drug only control.
  • the phenotypic effect of the delivery of drugs into cells was assessed via Vybrant fluorescent staining using flow cytometry to measure percentages of dead, apoptotic and live cells.
  • Act-D-Au-ZZ-hFtn(L29A L36A L81A L83A) (10% cell death) can be delivered into cells to release a payload of Act-D.
  • Compound nanocages composed of different types of subunit were also created by incubating the Au nanoparticle with His-ZZ-hFtn(L29A L36A L81A L83A) and His- GFP-hFtn(L29A L36A L81A L83A). Since the Au nanoparticle acts as the nucleating agent and the hFtn part of the fusion protein is identical, nanocages formed that contain the ZZ domain on some subunits and the GFP domain on others. These compound nanocages behaved as expected in terms of fluorescence and cellular delivery of drugs.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Nanotechnology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Inorganic Chemistry (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Dermatology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Medical Informatics (AREA)
  • Ceramic Engineering (AREA)
  • General Engineering & Computer Science (AREA)
  • Toxicology (AREA)
  • Optics & Photonics (AREA)
  • Zoology (AREA)
  • Radiology & Medical Imaging (AREA)

Abstract

L'invention concerne des nanocages, en particulier des nanocages de protéines, et spécifiquement des nanocages de ferritine. L'invention s'étend à des polypeptides de ferritine variants et à leurs acides nucléiques codants, à des nanocages de ferritine mutantes, ainsi qu'à leurs utilisations dans le diagnostic et l'administration de médicament, ainsi que dans des criblages phénotypiques dans le développement de médicaments.
EP17790822.5A 2016-10-20 2017-10-19 Nanocage Withdrawn EP3528849A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1617759.4A GB2555131A (en) 2016-10-20 2016-10-20 Nanocage
PCT/GB2017/053164 WO2018073593A1 (fr) 2016-10-20 2017-10-19 Nanocage

Publications (1)

Publication Number Publication Date
EP3528849A1 true EP3528849A1 (fr) 2019-08-28

Family

ID=57738302

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17790822.5A Withdrawn EP3528849A1 (fr) 2016-10-20 2017-10-19 Nanocage

Country Status (5)

Country Link
US (1) US20190240282A1 (fr)
EP (1) EP3528849A1 (fr)
JP (1) JP2019535246A (fr)
GB (1) GB2555131A (fr)
WO (1) WO2018073593A1 (fr)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111643684B (zh) * 2020-04-13 2021-05-04 中国科学院地质与地球物理研究所 一种具有肿瘤靶向性的钆基磁共振成像造影剂及制备方法
CN111631296B (zh) * 2020-05-08 2023-08-11 天津科技大学 一种以铁蛋白和橙皮素为原料构建食品功能因子传递系统的方法和应用
EP4161655A2 (fr) * 2020-06-08 2023-04-12 University of Washington Nanoparticules liées à un anticorps
US20230330033A1 (en) * 2020-06-08 2023-10-19 University Of Washington Designed antibody-bound nanoparticles
WO2022179536A1 (fr) * 2021-02-25 2022-09-01 昆山新蕴达生物科技有限公司 Mutant de sous-unité à chaîne lourde de ferritine et son application
CN117547618A (zh) * 2022-03-04 2024-02-13 南京纳么美科技有限公司 内腔装载小核酸药物的铁蛋白纳米笼载体及应用
CN114668683B (zh) * 2022-03-15 2023-09-08 杭州优玛达生物科技有限公司 一种纳米金掺杂自组装多肽活性物及其制备方法

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20020090546A (ko) * 2001-05-28 2002-12-05 주식회사 알엔에이 유산균 염색체 dna로의 삽입을 위한 삽입 벡터
KR101575194B1 (ko) * 2012-12-27 2015-12-07 경북대학교 산학협력단 인간 페리틴 유래 융합폴리펩티드

Also Published As

Publication number Publication date
GB201617759D0 (en) 2016-12-07
GB2555131A (en) 2018-04-25
US20190240282A1 (en) 2019-08-08
WO2018073593A1 (fr) 2018-04-26
JP2019535246A (ja) 2019-12-12

Similar Documents

Publication Publication Date Title
US20190240282A1 (en) Nanocage
Roncato et al. Improvement and extension of anti-EGFR targeting in breast cancer therapy by integration with the Avidin-Nucleic-Acid-Nano-Assemblies
Hu et al. Co-administration of iRGD with peptide HPRP-A1 to improve anticancer activity and membrane penetrability
JP7064769B2 (ja) 条件的活性型ポリペプチド
CA2993778C (fr) Proteine recombinee de la parkine a permeabilite cellulaire amelioree (icp) et son utilisation
JP2022513798A (ja) トランスフェリン受容体標的ペプチド
JP2022095641A (ja) 条件的活性型ポリペプチド及びそれを生成する方法
US20220098260A1 (en) BH4 Stabilized Peptides And Uses Thereof
KR20190062320A (ko) 글루타치온―s―전이효소 및 표적 세포 또는 표적 단백질 결합능을 갖는 단백질을 포함하는 융합 단백질 및 이의 용도
EP3481870A1 (fr) Anticorps humanisés franchissant la barrière hématoencéphalique et leurs utilisations
JP5677453B2 (ja) Bpbベースのカーゴ運搬システム
Tansi et al. New generation CPPs show distinct selectivity for cancer and noncancer cells
Ghanemi et al. Specific targeting of HER2-positive head and neck squamous cell carcinoma line HN5 by Idarubicin-ZHER2 affibody conjugate
Wöll et al. Sortase-A mediated chemoenzymatic lipidation of single-domain antibodies for cell membrane engineering
WO2021045210A1 (fr) Ferritine encapsulée dans un peptide
JP7282399B2 (ja) 断片化されたgrsポリペプチド、その変異体及びこれらの用途
KR101875809B1 (ko) 리피바디-항암 단백질 약물 복합체, 그 제조방법 및 용도
CA3158422A1 (fr) Peptides derives de la chromogranine a et leurs utilisations associees
JP7370598B2 (ja) ペプチド及びその使用
JP7429454B2 (ja) ペプチド及びその使用
Campeiro Potential applications of the cell-penetrating peptide crotamine
KR20240069453A (ko) 리소좀 표적화 접합체 및 이를 포함하는 암의 예방 또는 치료용 약학적 조성물
CN117203224A (zh) 包括封装在其中的客体货物的人工蛋白笼
CN109439637A (zh) 一种基于人钙调蛋白磷酸酶b亚基的靶向肽、制备方法及其应用

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20190423

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: IP2IPO INNOVATIONS LIMITED

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20191210